Potential Physiological Relevance of ERAD to the Biosynthesis of GPI-Anchored Proteins in Yeast - MDPI

Page created by Victoria Mcdaniel
 
CONTINUE READING
Potential Physiological Relevance of ERAD to the Biosynthesis of GPI-Anchored Proteins in Yeast - MDPI
International Journal of
               Molecular Sciences

Review
Potential Physiological Relevance of ERAD to the Biosynthesis
of GPI-Anchored Proteins in Yeast
Kunio Nakatsukasa

                                             Graduate School of Science, Nagoya City University, Yamanohata 1, Mizuho-cho, Mizuho-ku, Nagoya,
                                             Aichi 467-8501, Japan; nakatsukasa@nsc.nagoya-cu.ac.jp

                                             Abstract: Misfolded and/or unassembled secretory and membrane proteins in the endoplasmic
                                             reticulum (ER) may be retro-translocated into the cytoplasm, where they undergo ER-associated
                                             degradation, or ERAD. The mechanisms by which misfolded proteins are recognized and degraded
                                             through this pathway have been studied extensively; however, our understanding of the physio-
                                             logical role of ERAD remains limited. This review describes the biosynthesis and quality control of
                                             glycosylphosphatidylinositol (GPI)-anchored proteins and briefly summarizes the relevance of ERAD
                                             to these processes. While recent studies suggest that ERAD functions as a fail-safe mechanism for
                                             the degradation of misfolded GPI-anchored proteins, several pieces of evidence suggest an intimate
                                             interaction between ERAD and the biosynthesis of GPI-anchored proteins.

                                             Keywords: ERAD; GPI-anchored protein; Hrd1; Doa10; Ubc6; Ubc7; Ca2+ /Mn2+ P-type ATPase;
                                             Saccharomyces cerevisiae

                                   1. Introduction
         
                                                  Secretory and membrane proteins are translocated to the endoplasmic reticulum
Citation: Nakatsukasa, K. Potential          (ER), where they are folded into a three-dimensional conformation. In the ER, molecular
Physiological Relevance of ERAD to           chaperones recognize unfolded polypeptides and facilitate their folding by binding to
the Biosynthesis of GPI-Anchored
                                             amino acid patches containing exposed hydrophobic side chains [1]. Proteins that acquire
Proteins in Yeast. Int. J. Mol. Sci. 2021,
                                             the correct conformation are transported from the ER to the Golgi apparatus. However,
22, 1061. https://doi.org/10.3390/
                                             despite such protection, the maturation of proteins is an inherently error-prone process,
ijms22031061
                                             and misfolded proteins are frequently generated. To remove the potentially toxic misfolded
                                             proteins, eukaryotes have evolved two systems. The first system is the unfolded protein
Academic Editor: Jun Imai
                                             response (UPR), which involves the upregulation of the factors that increase the protein-
Received: 21 December 2020
Accepted: 19 January 2021
                                             folding capacity of the ER. The UPR also facilitates the transport of misfolded proteins
Published: 21 January 2021
                                             to the lysosome/vacuole, where they are degraded. The stress conditions caused by the
                                             accumulation of misfolded proteins (ER stress) induce membrane expansion of the ER to
Publisher’s Note: MDPI stays neutral
                                             alleviate the stress independently of an increase in ER chaperone levels [2,3]. The second
with regard to jurisdictional claims in
                                             system is ER-associated degradation (ERAD), by which terminally misfolded proteins
published maps and institutional affil-      are specifically recognized, retained in the ER, and retro-translocated to the cytoplasm,
iations.                                     where they are ubiquitinated and degraded by the proteasome. Components of the ERAD
                                             machinery can also be induced by the UPR [4–6]. Thus, the UPR and ERAD constitute
                                             two arms of the ER quality control apparatus and play critical roles in maintaining ER
                                             homeostasis [6–12].
Copyright: © 2021 by the author.
                                                  Various “model misfolded proteins” have been developed and used for the analysis of
Licensee MDPI, Basel, Switzerland.
                                             degradation pathways [13,14]. However, emerging evidence indicates that ERAD not only
This article is an open access article
                                             mediates the elimination of structurally abnormal proteins in the ER, but also contributes to
distributed under the terms and              the regulation of native proteins [15]. For example, ERAD targets properly folded proteins
conditions of the Creative Commons           to regulate metabolic enzymes, transcription factors, and metal transporters at the plasma
Attribution (CC BY) license (https://        membrane [16–19]. To further elucidate the physiological roles of ERAD, it is imperative to
creativecommons.org/licenses/by/             identify native substrates. In addition, yeast-based genetic interaction studies may help
4.0/).                                       discover novel associations between ERAD and other biological phenomena. This review

Int. J. Mol. Sci. 2021, 22, 1061. https://doi.org/10.3390/ijms22031061                                          https://www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2021, 22, 1061                                                                                           2 of 14

                                   describes the ERAD machineries in yeast and the findings of recent studies analyzing the
                                   biogenesis and quality control of misfolded glycosylphosphatidylinositol (GPI)-anchored
                                   proteins. The potential involvement of ERAD in manganese homeostasis, which might link
                                   the biogenesis of GPI-anchored proteins to ERAD, is also discussed.

                                   2. ER-Associated Degradation in Yeast
                                   2.1. The Hrd1 Pathway
                                        In yeast (S. cerevisiae), two dedicated ER membrane-associated E3 ligase complexes
                                   are involved in the recognition and degradation of misfolded proteins in the ER. The
                                   Hrd1 E3 ligase complex recognizes and targets misfolded luminal proteins, as well as
                                   ER membrane proteins with lesions at the transmembrane domain, for ubiquitination
                                   and degradation (Figure 1). These pathways are the so-called ERAD-L and -M pathways,
                                   respectively [17,20–27]. The Hrd1 complex consists of Hrd3, Usa1, Der1, Dfm1, Yos9,
                                   Kar2, Ubc7, and Cue1. Sucrose density gradient and systematic immunopurification
                                   analyses showed that Hrd1, Hrd3, Usa1, Der1, and Yos9 comprise the core complex [25,28].
                                   During ERAD, a misfolded substrate is first recognized by several factors, including Yos9
                                   (luminal lectin), Der1 (transmembrane protein), Kar2 (Hsp70 chaperone), or directly by
                                   Hrd1. The recognition mechanism for misfolded luminal glycoproteins has been studied
                                   extensively. The N-linked glycan is trimmed by glycosidase, and the terminal α1,6-mannose
                                   residue is recognized by the mannose 6-phosphate receptor homology (MRH) domain of
                                   Yos9 [29–33]. Non-glycosylated misfolded substrates may be recognized by Kar2. Both
                                   Yos9 and Kar2 bind to the luminal domain of Hrd3. In addition, unfolded and extended
                                   polypeptide segments may be recognized by the luminal domain of Hrd3 [34]. Until
                                   recently, differentiating the function of Hrd3 from Hrd1 stabilization was difficult because
                                   depletion of Hrd3 results in Hrd1 instability. However, removal of the ubiquitin-like
                                   domain of Usa1 caused Hrd1 to remain stable. This method was used to demonstrate
                                   that Hrd3 plays a direct role in facilitating the transfer of ubiquitin to substrates [35].
                                   Usa1 mediates the interaction of Hrd1 with Der1, which is an inactive form of rhomboid
                                   protease, and transports substrates from the ER lumen to Hrd1 for degradation. Usa1 can
                                   also mediate the formation of an Hrd1 oligomer, which is critical for the degradation of
                                   ERAD-M substrates [21,27,36]. After recognition, the substrate is transferred to the Hrd1
                                   complex for polyubiquitination. The transmembrane protein Cue1 is an ER membrane
                                   protein that recruits the E2 ubiquitin-conjugating enzyme Ubc7 to the Hrd1 complex [37].
                                   Hrd1 also recruits the ER membrane protein Ubx2 to the complex, which anchors the
                                   AAA+ ATPase Cdc48/p97 to the membrane [38,39]. Then, Cdc48/p97 hydrolyzes ATP and
                                   liberates the substrate from the ER. Inactivation of Cdc48 leads to a formation of stalled
                                   retro-translocation complex containing Hrd1, Usa1, Hrd3, Der1, the 26S proteasome, Yos9,
                                   ubiquitinated substrates, and Cdc48. This suggests that substrate recognition and retro-
                                   translocation might be coupled, at least for some substrates [40]. Recognition of the integral
                                   membrane ERAD-M substrate is mediated by the transmembrane domain of Hrd1 [41]
                                   (Figure 1). The membrane substrates may exit the ER through a distinct pathway mediated
                                   by the Dfm1 rhomboid protein, which can also recruit the AAA+ ATPase Cdc48/p97 to the
                                   membrane for retro-translocation. Upon deletion of the DFM1 gene, Hrd1 levels increase
                                   and the Hrd1 complex may be remodeled, thereby enabling a novel route of membrane
                                   protein retro-translocation. This supports the functional flexibility of the Hrd1 complex in
                                   response to ER stress [42,43].

                                   2.2. The Doa10 Pathway
                                        Another E3 ligase complex involved in yeast ERAD is Doa10, which resides in the
                                   nuclear membrane and ER membrane. Doa10 is a 150 kDa protein with 14 transmembrane
                                   segments [44] that targets transmembrane substrates with cytoplasmic lesions for ubiquiti-
                                   nation and degradation, a pathway called ERAD-C (Figure 1). Substrates with abnormal
                                   structural domains in multiple regions may be degraded depending on both the Hrd1
                                   and Doa10 complexes [45,46]. Substrates of the Doa10 complex include single- or multi-
2.2. The Doa10 Pathway
                                         Another E3 ligase complex involved in yeast ERAD is Doa10, which resides in the
                                    nuclear membrane and ER membrane. Doa10 is a 150 kDa protein with 14 transmembrane
                                    segments [44] that targets transmembrane substrates with cytoplasmic lesions for ubiqui-
Int. J. Mol. Sci. 2021, 22, 1061    tination and degradation, a pathway called ERAD-C (Figure 1). Substrates with abnormal                      3 of 14
                                    structural domains in multiple regions may be degraded depending on both the Hrd1 and
                                    Doa10 complexes [45,46]. Substrates of the Doa10 complex include single- or multi-span-
                                    spanning
                                    ning        membrane
                                          membrane            proteins
                                                         proteins         in the
                                                                     in the   ER ER andand
                                                                                         thethe   inner
                                                                                               inner      nuclearmembrane,
                                                                                                       nuclear      membrane,asaswellwellasas soluble
                                                                                                                                              soluble
                                    proteins in the cytosol and nucleoplasm [19,44,47–52]. Unlike the Hrd1 complex, which
                                    proteins
                                    comprises multiple
                                    comprises    multiplecomponents,
                                                              components,the     theDoa10
                                                                                       Doa10complex
                                                                                                complex  is relatively simple
                                                                                                             is relatively      and contains
                                                                                                                            simple              three
                                                                                                                                      and contains
                                    ubiquitination
                                    three             enzymes:
                                           ubiquitination           Ubc6, Ubc7,
                                                              enzymes:      Ubc6, and     Cue1.
                                                                                      Ubc7,   andHowever,       in contrast
                                                                                                     Cue1. However,          to the Hrd1
                                                                                                                         in contrast        pathway,
                                                                                                                                        to the  Hrd1
                                    the mechanism
                                    pathway,            by which by
                                                the mechanism        thewhich
                                                                          Doa10the   complex
                                                                                       Doa10 recognizes         misfolded
                                                                                                complex recognizes          substrates
                                                                                                                          misfolded       is not well
                                                                                                                                        substrates   is
                                    understood.     Cytosolic    chaperones,       such  as  the   Hsp70    Ssa1  and
                                    not well understood. Cytosolic chaperones, such as the Hsp70 Ssa1 and the cytosolicthe cytosolic   Hsp40s    Ydj1
                                    and Hlj1,
                                    Hsp40s      facilitate
                                             Ydj1           substrate
                                                    and Hlj1,            recognition
                                                                 facilitate  substrate[53–55].
                                                                                          recognition The [53–55].
                                                                                                           degrons The
                                                                                                                     of Doa10
                                                                                                                          degronssubstrates
                                                                                                                                     of Doa10  can  be
                                                                                                                                                 sub-
                                    cytoplasmic     [49,50]   or  located    within    the TM      region   [51].  During   ubiquitination,
                                    strates can be cytoplasmic [49,50] or located within the TM region [51]. During ubiquiti-                   Ubc6
                                    attachesUbc6
                                    nation,    the first ubiquitin
                                                    attaches          to aubiquitin
                                                                the first   substrate,toand     Ubc7 extends
                                                                                           a substrate,            ubiquitin
                                                                                                            and Ubc7          chains
                                                                                                                        extends        using mostly
                                                                                                                                   ubiquitin   chains
                                    lysine mostly
                                    using  48 linkages    [56].
                                                     lysine  48 Ubiquitinated
                                                                 linkages [56]. substrates
                                                                                     Ubiquitinated are then  retro-translocated
                                                                                                        substrates                 to the cytosol by
                                                                                                                     are then retro-translocated
                                    Cdc48/p97,
                                    to the cytosolwhich     is recruited
                                                     by Cdc48/p97,          to the
                                                                        which        ER membrane
                                                                                  is recruited    to thebyERUbx2    and/orby
                                                                                                              membrane       Dfm1
                                                                                                                               Ubx2[43,53,57].
                                                                                                                                       and/or Dfm1The
                                    mechanism      underlying      the retro-translocation         of membrane      substrates,
                                    [43,53,57]. The mechanism underlying the retro-translocation of membrane substrates, in-      including    Doa10
                                    substrates,
                                    cluding      is ill-defined.
                                              Doa10    substrates,  The
                                                                      is extraction
                                                                         ill-defined.ofThe Ubc6,    which isofdegraded
                                                                                                extraction                 in a Doa10-dependent
                                                                                                                  Ubc6, which     is degraded in a
                                    manner     [50,58],  was   recently    reconstituted       in  vitro  [52].
                                    Doa10-dependent manner [50,58], was recently reconstituted in vitro [52].    The  results  of this
                                                                                                                                     The experiment
                                                                                                                                           results of
                                    this experiment suggest that the luminal domain is unfolded by the action of cytosol
                                    suggest   that the  luminal    domain     is  unfolded    by   the action   of Cdc48/p97     in the           and
                                                                                                                                          Cdc48/p97
                                    crosses  the  membrane       in an  unfolded      state.
                                    in the cytosol and crosses the membrane in an unfolded state.

                 Figure
                 Figure 1.
                        1. The
                           The ERAD
                               ERAD (ER-associated
                                    (ER-associated degradation)
                                                   degradation) pathway
                                                                pathway in
                                                                        in Saccharomyces
                                                                           Saccharomyces cerevisiae (see text
                                                                                         cerevisiae (see text for
                                                                                                              for detail).
                                                                                                                  detail).

                                    3. GPI-Anchored Proteins
                                    3. GPI-Anchored Proteins
                                          GPI anchors are structurally complex glycophospholipids that are post-translation-
                                          GPI anchors are structurally complex glycophospholipids that are post-translationally
                                    ally attached to the C-terminus of secretory proteins (Figure 2). The highly conserved core
                                    attached to the C-terminus of secretory proteins (Figure 2). The highly conserved core
                                    structure of the GPI anchor precursor (CP2: complete precursor 2), which accumulates in
                                    structure of the GPI anchor precursor (CP2: complete precursor 2), which accumulates in
                                    the GPI transamidase mutant, comprises four mannoses (Man1, Man2, Man3, and Man4),
                                    the GPI transamidase mutant, comprises four mannoses (Man1, Man2, Man3, and Man4),
                                    three ethanolamine phosphate (EtN-P) substituents on Man1, Man2, and Man3, one acyl-
                                    three ethanolamine phosphate (EtN-P) substituents on Man1, Man2, and Man3, one acyl-
                                    phosphatidylinositol (acyl-PI), and one glucosamine (GlcN) [59]. More than 20 genes en-
                                    phosphatidylinositol (acyl-PI), and one glucosamine (GlcN) [59]. More than 20 genes
                                    coding enzymes involved in the biosynthesis of GPI anchors have been identified by ge-
                                    encoding enzymes involved in the biosynthesis of GPI anchors have been identified by
                                    netic screening to isolate mutant cells that lack GPI proteins at the surface. GPI anchor
                                    genetic screening to isolate mutant cells that lack GPI proteins at the surface. GPI anchor
                                    synthesis begins on the cytosolic side of the ER membrane. The first step in GPI biosyn-
                                    thesis is the addition of N-Acetylglucosamine (GlcNAc) to phosphatidylinositol (PI). This
                                    transfer reaction is catalyzed by GPI-GlcNAc transferase, which consists of Gpi15, Eri1,
                                    Gpi3, Gpi19, Gpi2, and Gpi1 [60–62]. Next, the acetyl group of GlcNAc is removed by
                                    Gpi12, a GlcNAc-PI de-N-acetylase, to generate GlcN-PI, which is translocated by flippase
                                    to the luminal side of the ER membrane. Inside the ER, the acyltransferase Gwt1 adds an
synthesis begins on the cytosolic side of the ER membrane. The first step in GPI biosyn-
                                   thesis is the addition of N-Acetylglucosamine (GlcNAc) to phosphatidylinositol (PI). This
                                   transfer reaction is catalyzed by GPI-GlcNAc transferase, which consists of Gpi15, Eri1,
Int. J. Mol. Sci. 2021, 22, 1061   Gpi3, Gpi19, Gpi2, and Gpi1 [60–62]. Next, the acetyl group of GlcNAc is removed                  4 ofby
                                                                                                                                          14
                                   Gpi12, a GlcNAc-PI de-N-acetylase, to generate GlcN-PI, which is translocated by flippase
                                   to the luminal side of the ER membrane. Inside the ER, the acyltransferase Gwt1 adds an
                                   acyl chain,
                                   acyl chain, which
                                                which is
                                                       is mostly
                                                          mostlypalmitate,
                                                                  palmitate,totothe
                                                                                 the2-position
                                                                                     2-positionofofthe
                                                                                                     theinositol
                                                                                                          inositolinin
                                                                                                                     GlcN-PI,
                                                                                                                       GlcN-PI,generating
                                                                                                                                 generat-
                                   GlcN-(acyl)   PI [63]. Subsequently,   Man1   and  Man2   are transferred     from
                                   ing GlcN-(acyl) PI [63]. Subsequently, Man1 and Man2 are transferred from dolichol- dolicholphospho-
                                   mannose (Dol-P-Man)
                                   phosphomannose             to GlcN-(acyl)
                                                        (Dol-P-Man)           PI by GPIPImannosyl-transferase
                                                                       to GlcN-(acyl)      by GPI mannosyl-transferase1 (Gpi14)1and    GPI
                                                                                                                                  (Gpi14)
                                   mannosyl-transferase     2 (Gpi18), respectively  [64,65]. In addition,    EtN-P   is transferred
                                   and GPI mannosyl-transferase 2 (Gpi18), respectively [64,65]. In addition, EtN-P is trans-         from
                                   phosphatidylethanolamine
                                   ferred                         (PE) to Man1(PE)
                                           from phosphatidylethanolamine          by EtN-P  transferase
                                                                                      to Man1    by EtN-P   (Mcd4)   [66]. Then,
                                                                                                               transferase        Man3
                                                                                                                             (Mcd4)       is
                                                                                                                                      [66].
                                   added to Man2 by GPI mannosyl-transferase 3 (Gpi10), and Man4 is added to Man3 by
                                   Then, Man3 is added to Man2 by GPI mannosyl-transferase 3 (Gpi10), and Man4 is added
                                   Smp3 before the addition of EtN-P to Man3 by EtN-P transferase 3 [67–69]. This complex
                                   to Man3 by Smp3 before the addition of EtN-P to Man3 by EtN-P transferase 3 [67–69].
                                   comprises Gpi13, which is a catalytic subunit, and the stabilizing subunit Gpi11. Finally,
                                   This complex comprises Gpi13, which is a catalytic subunit, and the stabilizing subunit
                                   EtN-P is added to Man2 by EtN-P transferase enzyme 2, which consists of the catalytic
                                   Gpi11. Finally, EtN-P is added to Man2 by EtN-P transferase enzyme 2, which consists of
                                   subunit Gpi7 and the stabilizing subunit Gpi11 [70,71].
                                   the catalytic subunit Gpi7 and the stabilizing subunit Gpi11 [70,71].

        Figure2.2.The
       Figure      The  pathway
                      pathway of of
                                 GPIGPI (glycosylphosphatidylinositol)
                                     (glycosylphosphatidylinositol)    biosynthesis
                                                                    biosynthesis in in Saccharomyces
                                                                                    Saccharomyces    cerevisiae
                                                                                                  cerevisiae    (see
                                                                                                             (see texttext
                                                                                                                        forfor de-
                                                                                                                            detail).
                                                               tail).
                                         Precursors of GPI-anchored proteins have a signal for GPI anchoring at the C-terminus
                                   and Precursors   of GPI-anchored
                                        a conventional   signal sequenceproteins
                                                                             for have  a signal for GPI
                                                                                  ER translocation        anchoring
                                                                                                      at the          at the The
                                                                                                              N-terminus.    C-termi-
                                                                                                                                  GPI
                                   nus and  a conventional  signal  sequence    for ER  translocation  at the N-terminus.
                                   anchoring sequence contains a C-terminal hydrophobic domain that is separated from        The  GPI
                                   anchoring  sequence
                                   the upstream         contains a C-terminal
                                                  GPI-attachment     site (the “ωhydrophobic     domain
                                                                                   site”) by a short      thatof
                                                                                                      stretch  is hydrophilic
                                                                                                                  separated from   the
                                                                                                                                amino
                                   upstream
                                   acids [72].GPI-attachment   site (the
                                              Soon after translation      “ω precursor
                                                                      of the site”) by aprotein
                                                                                          short stretch of hydrophilic
                                                                                                 by ribosomes             amino acids
                                                                                                                 and its translocation
                                   into the ER membrane are completed, the C-terminus of the protein is conjugated to the
                                   amine group of EtN-P by the GPI transamidase complex, which consists of five essential
                                   proteins: Gpi8, Gpi17, Gpi16, Gab1, and Gaa1 [73–77]. Of these, Gpi8, a catalytic subunit
                                   that is homologous to caspase-like cysteine proteases, cleaves the C-terminus of substrate
                                   proteins. This reaction is a prerequisite for the transamidation reaction [78–80]. After
                                   attachment, the GPI anchor is subject to a sequence of remodeling reactions on both the
Int. J. Mol. Sci. 2021, 22, 1061                                                                                           5 of 14

                                   lipid and sugar moieties. These reactions occur exclusively inside the ER and are catalyzed
                                   by Bst1, Per1, Gup1, and Cwh43 in yeast [81–84]. Bst1 is a phosphatidylinositol deacylase
                                   that mediates inositol deacylation. This step is required for downstream lipid remodeling.
                                   Per1 removes the unsaturated fatty acid at the sn2 position, and Gup1 adds C26 fatty
                                   acids. Cwh43 is responsible for replacing diacylglycerol (DAG) with ceramide, which is a
                                   major lipid (saturated and very long inositolphosphoceramide) component of mature GPI
                                   anchors in yeast. Subsequently, Cdc1 and Ted1 remove the EtN-P of the first and second
                                   mannose, respectively [85,86]. These two enzymes are homologs of mammalian PGAP5,
                                   a membrane-spanning enzyme that possesses a metal-containing phosphoesterase motif
                                   in the luminal domain and removes EtN-P from the second mannose in the ER [87]. The
                                   removal of EtN-P from the second mannose is a prerequisite for the recognition of GPI-
                                   anchored proteins by the p24 complex and their exit from the ER. The localization of Cdc1
                                   to the cis/medial Golgi apparatus, and not to the ER, was demonstrated recently. Removal
                                   of EtN-P from the second mannose by Ted1 in the ER and from the first mannose by Cdc1
                                   in the Golgi apparatus may serve as a quality assurance signal for GPI-anchored proteins.

                                   4. Quality Control of GPI-Anchored Proteins
                                         In yeast, a mutant version of Gas1, β-1,3-glucanosyltransferase, which is referred to as
                                   Gas1*, is a well-studied model substrate for the quality control of misfolded GPI-anchored
                                   proteins. Gas1* contains a point mutation (G291R) that renders the protein misfolded
                                   and leads to its degradation [88,89]. Accumulating evidence suggests that Gas1* is not an
                                   efficient ERAD substrate. Only a small fraction is delivered to the ERAD pathway, while the
                                   vast majority of proteins escape Hrd1-dependent degradation; however, they are rapidly
                                   recognized by the p24 complex, including Emp24, exported from the ER, and delivered to
                                   the vacuole for degradation. p24 family proteins are conserved transmembrane proteins of
                                   ~24 kDa that function as cargo receptors for GPI-anchored proteins. The budding yeast p24
                                   family is composed of three p24α (Erp1, Erp5, Erp6), one p24β (Emp24), three p24γ (Erp2,
                                   Erp3, Erp4), and one p24δ (Erv25) [90]. These are single-transmembrane proteins with a
                                   short (10–20 amino acids) C-terminal cytoplasmic tail. The cytoplasmic tail can interact
                                   with both COPI (Coat Protein I) and COPII (Coat Protein II) subunits. The luminal portion
                                   of these proteins may contribute to the formation of p24 oligomer and also participate in the
                                   cargo recognition. In mammalian cells, the misfolded version of the prion protein (PrP*),
                                   which is a GPI-anchored protein, is not degraded by ERAD but rather exported from the
                                   ER despite the misfolding [91]. PrP* is recognized by the p24 complex and delivered to the
                                   plasma membrane, from where it is transported to the lysosome for degradation [92]. When
                                   the ER is loaded with high amounts of misfolded proteins and its capacity is saturated
                                   during ER stress, misfolded PrP* dissociates from resident ER chaperones and is rapidly
                                   released into the secretory pathway in a process called rapid ER stress-induced export
                                   (RESET) [92]. This response is faster than the activation of the UPR and reduces the load of
                                   aberrant proteins in the ER, thereby maintaining protein homeostasis in the ER.
                                         Then, what is the difference between misfolded GPI-anchored proteins and general
                                   ERAD substrates? When GPI anchor attachment is impaired, the misfolded protein moiety,
                                   which is free from the ER membrane, is rarely targeted to the vacuole. Instead, a consider-
                                   able portion of these proteins is delivered to the ERAD pathway. The protein moiety of
                                   Gas1* is delivered to the Hrd1-dependent degradation pathway. In mammals, when GPI
                                   anchor attachment is prevented, PrP* is targeted for ERAD. Therefore, the protein moiety of
                                   the misfolded GPI-anchored proteins can be disposed by ERAD. It is possible that the GPI
                                   anchor sterically obstructs step(s) during ERAD, including retro-translocation and/or other
                                   steps. However, when GPI anchor remodeling proceeds incorrectly in cells lacking Bst1,
                                   Cwh43, or Ted1, GPI-anchored proteins are not efficiently exported from the ER; instead,
                                   they are retained in the ER and delivered to the Hrd1-dependent ERAD pathway [93].
                                   This observation supports the idea that a GPI anchor does not pose a steric obstruction to
                                   the ERAD of misfolded GPI-anchored proteins. The ER retention time of misfolded GPI-
                                   anchored proteins may be determined by the remodeling status of the GPI anchor, which is
Int. J. Mol. Sci. 2021, 22, 1061                                                                                           6 of 14

                                   directly coupled to ER export. To ensure the correct sequential synthesis of GPI-anchored
                                   proteins, it would be beneficial to remove those possessing an immature GPI anchor from
                                   the ER. Thus, the quality control of GPI-anchored proteins may be more severe than that of
                                   normal secretory proteins. This idea is further supported by the recent observation that
                                   Cdc1, a remodeling enzyme for GPI-anchored proteins, resides in the cis/medial Golgi
                                   apparatus, where additional quality control systems might be required [94]. In addition, a
                                   recent study showed that PrP* is trafficked from the ER to lysosomes in a complex with
                                   ER-derived chaperones, including calnexin and cargo receptors [95]. These interaction
                                   partners are critical for rapid endocytosis. Resident ER factors not only protect misfolded
                                   GPI-anchored proteins from aggregation during trafficking, but also ensure that they are
                                   subject to quality control at the plasma membrane and endocytosis to lysosomes.

                                   5. Potential Physiological Relevance of ERAD to the Biosynthesis of
                                   GPI-Anchored Proteins
                                        As mentioned above, emerging evidence suggests that ERAD functions as a fail-
                                   safe mechanism for the degradation of misfolded GPI-anchored proteins when the vac-
                                   uole/lysosomal route is impaired. However, several observations support the physiological
                                   relevance of ERAD to the biosynthesis of GPI-anchored proteins.

                                   5.1. Genetic Interactions between GPI and ERAD Genes
                                        Systematic studies in yeast have suggested several positive and negative genetic
                                   interactions between genes encoding ERAD components and GPI biosynthetic factors [96].
                                   Negative genetic interactions refer to double mutants that exhibit a more severe fitness
                                   defect than expected [97]. Conversely, positive genetic interactions refer to double mutants
                                   with a less severe growth defect than anticipated. These genes may contain genes encoding
                                   components of the same nonessential protein complex [97]. Genes required for relatively
                                   the later steps of GPI biosynthesis tend to interact with ERAD genes. For example, HRD1
                                   shows positive genetic interactions with GPI8, GPI10, GPI11, and GPI17. Similarly, HRD3
                                   shows positive genetic interactions with GPI8, GPI10, and GPI17, and negative genetic
                                   interactions with GPI19. UBC7 shows positive genetic interactions with GPI13, GPI16, and
                                   GPI17 [96]. Other interactions between major ERAD components and factors involved
                                   in the biosynthesis of GPI-anchored proteins are listed in Table 1. Although the reasons
                                   for these genetic interactions are currently unknown, one possible explanation is that
                                   endogenous substrates that accumulate upon ERAD deficiency positively or negatively
                                   affect the GPI deletion phenotype.

                                   Table 1. Positive and negative genetic interactions between genes encoding ERAD components and
                                   GPI biosynthetic factors. N: negative genetic interactions; P: positive genetic interactions.

                                                GPI2      GPI8      GPI10     GPI11      GPI13     GPI16      GPI17     GPI19
                                    HRD1                    P         P          P                              P
                                    HRD3                    P         P                                         P         N
                                    UBC7                                                   P          P         P
                                    USA1                    P                    P         P                    P
                                    DER1         N                               P         P
                                    YOS9                                                   P                    N         N
                                    DOA10                                        P

                                        In mammals, genome-wide CRISPR-Cas9 (Clustered Regularly Interspaced Short
                                   Palindromic Repeats/CRISPR-Associated Proteins 9) genetic screening suggests that dis-
                                   ruption of HRD1 or several other ERAD components enhances GPI synthesis in GPI-
                                   transamidase-deficient cells [98]. The proposed scenario is that cells use ERAD to suppress
                                   GPI synthesis by degrading unknown protein factor(s) or endogenous substrate(s) that
                                   normally enhance the biosynthesis of GPI. Disruption of ERAD may cause the accumu-
Int. J. Mol. Sci. 2021, 22, 1061                                                                                          7 of 14

                                   lation of such factor(s), which can lead to increased free GPIs including its biosynthetic
                                   intermediates as well as mature forms in GPI-transamidase-deficient cells [98].

                                   5.2. Quality Control of Proteins that Harbor the GPI Anchoring Signal in the Cytosol
                                        Molecular recognition events, including protein targeting to the organelle, are inher-
                                   ently imperfect because of intrinsic limits on specific binding. Indeed, during protein
                                   targeting to the ER, many secretory proteins fail to associate with the signal recognition
                                   particle (SRP) and can be detected in the cytosol before their translocation [99,100]. Under
                                   normal conditions in mammalian cells, the efficiency of ER translocation is not high: it
                                   may range from 60% to 95% [101]. This implies that the cell must be equipped with a
                                   quality control system to monitor a significant number of un-translocated proteins in the
                                   cytosol. The existence of a surveillance system that targets SRP-independent substrates
                                   for degradation is also likely. These would include a precursor form of GPI-anchored
                                   proteins whose C-terminal signal is hydrophobic. In yeast, GPI-anchored proteins that are
                                   not translocated to the ER are degraded on the cytosolic face of the ER. This degradation
                                   pathway is termed prERAD (Pre-insertional ERAD) and relies on opposing forces of the
                                   ubiquitin ligase Doa10 and the deubiquitinating enzyme Ubp1 [102].

                                   5.3. Exit of GPI-Anchored Proteins from the ER Is Affected by the Perturbation of
                                   Manganese Homeostasis
                                         Studies of mammalian cells show that PGAP5, an ER membrane protein, has a metal-
                                   containing phosphate esterase motif in the lumen and requires manganese ions (Mn2+ )
                                   for its enzymatic activity [87]. PGAP5 catalyzes the removal of the second EtN-P after
                                   GPI is transferred to the protein. The removal of EtN-P from GPI-glycan by PGAP5 is
                                   required for the efficient transport of GPI-anchored proteins from the ER to the Golgi
                                   apparatus. As mentioned above, S. cerevisiae has two putative homologs of PGAP5, Cdc1
                                   and Ted1. Cdc1 is a Mn2+ -dependent enzyme that interacts with genes involved in GPI
                                   fatty acid remodeling [85,103]. Cdc1 was recently shown to have mannose-ethanolamine
                                   phosphate phosphodiesterase activity, and it is responsible for the removal of EtN-P from
                                   Man1 [85]. Transport of Gas1 from the ER is delayed in ted1∆ cells [104,105], although it
                                   is not currently clear whether Ted1 is a manganese-dependent enzyme. Perturbation of
                                   manganese homeostasis by depletion of Spf1, a P5-type ATPase that regulates manganese
                                   transport into the ER, causes the defective export of GPI-anchored proteins from the ER,
                                   and their accumulation in the ER [106]. These observations in mammals and yeast suggest
                                   that manganese homeostasis is critical for the biosynthesis of GPI-anchored proteins.

                                   5.4. Possible Involvement of ERAD in the Maintenance of Manganese Homeostasis
                                         I found a genetic interaction between ERAD and Pmr1, a P-type Ca2+ - and Mn2+ -
                                   transporting ATPase that is localized in the Golgi membrane [107], in yeast. Pmr1 is a
                                   prototypic member of the Ca2+ -ATPase family of transporting ATPases, which are found in
                                   a variety of organisms, including fungi, Caenorhabditis elegans, Drosophila melanogaster, and
                                   mammals [108–110]. Both the calcium and manganese ions that are transported by Pmr1
                                   into the Golgi lumen enable proper processing and trafficking of proteins through the secre-
                                   tory pathway [107,111–113]. While calcium is mainly required for protein trafficking [114],
                                   manganese is an essential enzymatic co-factor for glycosyltransferases that catalyze protein
                                   glycosylation in the secretory pathway [112,115]. Pmr1 transports excess cytosolic man-
                                   ganese into the Golgi lumen and mediates its export from the cell via secretory pathway
                                   vesicles. Thus, Pmr1 contributes to the cellular detoxification of manganese [115]. Yeast
                                   cells with a deleted PMR1 gene (pmr1∆ cells) show a pleiotropic phenotype. For example,
                                   cells lacking Pmr1 are sensitive to ethylene glycol-bis(β-aminoethyl ether)-N,N,N0 ,N0 -
                                   tetraacetic acid (EGTA), an effective chelator of calcium, manganese, and other divalent
                                   metal ions [107]. Depletion of Pmr1 results in the accumulation of manganese and calcium
                                   in the cytoplasm as well as depletion of these ions from the Golgi apparatus. Moreover,
                                   cells lacking Pmr1 display a defect in carboxy peptidase Y (CPY) trafficking [112] and a
                                   defect in the degradation of CPY*, a typical ERAD-L substrate [116]. Defects in the human
Yeast cells with a deleted PMR1 gene (pmr1∆ cells) show a pleiotropic phenotype. For
                                    example, cells lacking Pmr1 are sensitive to ethylene glycol-bis(β-aminoethyl ether)-
                                    N,N,N′,N′-tetraacetic acid (EGTA), an effective chelator of calcium, manganese, and other
                                    divalent metal ions [107]. Depletion of Pmr1 results in the accumulation of manganese
Int. J. Mol. Sci. 2021, 22, 1061    and calcium in the cytoplasm as well as depletion of these ions from the Golgi apparatus.                   8 of 14
                                    Moreover, cells lacking Pmr1 display a defect in carboxy peptidase Y (CPY) trafficking
                                    [112] and a defect in the degradation of CPY*, a typical ERAD-L substrate [116]. Defects
                                    in the human
                                   ortholog    of PMR1,ortholog
                                                             ATP2C1,of PMR1,    ATP2C1, are
                                                                           are associated        associated
                                                                                              with             with Hailey–Hailey
                                                                                                    Hailey–Hailey                         disease, an
                                                                                                                         disease, an autosomal
                                    autosomalblistering
                                   dominant       dominant       blistering
                                                              skin   disorder skin  disorder [108,117].
                                                                                [108,117].
                                         AAprevious
                                              previouslarge-scale
                                                          large-scaleanalysis
                                                                         analysissuggested
                                                                                     suggestedaagenetic
                                                                                                     geneticinteraction       betweenPMR1
                                                                                                               interactionbetween         PMR1and  and
                                   UBC7
                                    UBC7[118].
                                            [118]. To confirm this  this observation,
                                                                          observation,I Iconstructed        ubc7∆,
                                                                                            constructedubc7∆,         pmr1∆,
                                                                                                                   pmr1∆,    andand   ubc7∆pmr1∆
                                                                                                                                  ubc7∆pmr1∆       mu-
                                   mutant    strains
                                    tant strains   andand    analyzed
                                                         analyzed         their
                                                                       their     growth
                                                                             growth        (Supplementary
                                                                                       (Supplementary           Materials).
                                                                                                            Materials).        However,
                                                                                                                           However,          double
                                                                                                                                         double    mu-
                                   mutant    cells
                                    tant cells      grewnormally
                                                  grew      normallycompared
                                                                         comparedwith  with isogenic
                                                                                              isogenic wild-type
                                                                                                         wild-type strains or    or single
                                                                                                                                     singlemutant
                                                                                                                                              mutant
                                   strains,
                                    strains,atatleast
                                                 leastononyeast
                                                            yeastextract-peptone-dextrose
                                                                    extract-peptone-dextrose(YPD)   (YPD)medium
                                                                                                            medium(Figure(Figure3).
                                                                                                                                  3).Next,
                                                                                                                                       Next,I Itested
                                                                                                                                                tested
                                   the  EGTA     sensitivity    of  these   strains  because    pmr1∆
                                    the EGTA sensitivity of these strains because pmr1∆ cells were previously showntotobe
                                                                                                         cells  were   previously     shown           be
                                   sensitive
                                    sensitivetotoEGTA.
                                                    EGTA.Consistently,
                                                              Consistently,  pmr1∆
                                                                                pmr1∆ cells were
                                                                                         cells    sensitive
                                                                                               were          to EGTA,
                                                                                                      sensitive   to EGTA,whereas
                                                                                                                               whereascellscells
                                                                                                                                            withwith
                                                                                                                                                   the
                                   deleted   UBC7UBC7
                                    the deleted       genegene
                                                             grewgrewsimilarly   to wild-type
                                                                           similarly             cells (Figure
                                                                                       to wild-type              3). Interestingly,
                                                                                                       cells (Figure                   I found
                                                                                                                        3). Interestingly,        that
                                                                                                                                             I found
                                   the
                                    thatdeletion   of UBC7
                                          the deletion          suppressed
                                                          of UBC7              the EGTA
                                                                       suppressed           sensitivity
                                                                                      the EGTA            of pmr1∆
                                                                                                   sensitivity         cells,cells,
                                                                                                                 of pmr1∆     indicating    that the
                                                                                                                                     indicating    that
                                   deletion   of  UBC7    suppresses      the  EGTA    sensitivity   of pmr1∆    cells.  To
                                    the deletion of UBC7 suppresses the EGTA sensitivity of pmr1∆ cells. To further confirm  further   confirm    this
                                   result,  I analyzed
                                    this result,           the genetic
                                                   I analyzed              interactions
                                                                   the genetic             between
                                                                                  interactions        PMR1PMR1
                                                                                                  between      and other     ERADERAD
                                                                                                                       and other      components.
                                                                                                                                             compo-
                                   As            in  Figure   3,  the EGTA     sensitivity   of pmr1∆
                                    nents. As shown in Figure 3, the EGTA sensitivity of pmr1∆ cells was rescued depletion
                                       shown                                                             cells  was  rescued    by   the   by the de-
                                   of Hrd1, of
                                    pletion    a retro-translocation       channel for
                                                  Hrd1, a retro-translocation             luminalfor
                                                                                        channel     ERAD     substrates.
                                                                                                       luminal               A similar trend
                                                                                                                  ERAD substrates.                was
                                                                                                                                           A similar
                                   observed     for  Hrd3,    a  component      of  the  Hrd1   complex,    and   for
                                    trend was observed for Hrd3, a component of the Hrd1 complex, and for Doa10, a mem- Doa10,   a  membrane        E3
                                   ligase
                                    branethat    targets
                                            E3 ligase      misfolded
                                                        that             membrane
                                                              targets misfolded          proteins with
                                                                                      membrane            cytoplasmic
                                                                                                   proteins                lesions. These
                                                                                                               with cytoplasmic              results
                                                                                                                                      lesions.  These
                                   suggest    a potential    physiological      role  of  ERAD    in maintaining      calcium
                                    results suggest a potential physiological role of ERAD in maintaining calcium and man-       and    manganese
                                   homeostasis      in the Golgi
                                    ganese homeostasis         in theapparatus.
                                                                        Golgi apparatus.

                                    Figure3.
                                   Figure  3. Deletion
                                               Deletionofofmajor
                                                            majorERAD
                                                                   ERADcomponents
                                                                           componentssuppresses   the EGTA
                                                                                          suppresses  the EGTA(Ethylene  glycol-bis(β-ami-
                                                                                                                    (Ethylene  glycol-bis(β-
                                    noethyl  ether)-N,N,N′,N′-tetra-acetic acid) sensitivity of pmr1∆ cells. Serial 10-fold dilutions
                                   aminoethyl ether)-N,N,N0 ,N0 -tetra-acetic acid) sensitivity of pmr1∆ cells. Serial 10-fold        of of
                                                                                                                                dilutions
                                    yeast cultures were spotted onto yeast extract-peptone-dextrose medium (YPD) or YPD containing
                                   yeast cultures were spotted onto yeast extract-peptone-dextrose medium (YPD) or YPD containing
                                    2 mM EGTA. Plates were incubated for 3–7 days. The strains used here were BY4741 (wild-type)
                                   2and
                                     mMitsEGTA.
                                           mutantPlates were incubated for 3–7 days. The strains used here were BY4741 (wild-type) and
                                                    derivatives.
                                   its mutant derivatives.
                                          The mechanism by which depletion of ERAD might rescue the EGTA-sensitive phe-
                                         The mechanism by which depletion of ERAD might rescue the EGTA-sensitive pheno-
                                    notype of pmr1∆ is currently unclear. One possible scenario is that a native luminal and/or
                                   type of pmr1∆ is currently unclear. One possible scenario is that a native luminal and/or
                                    membrane protein(s) accumulates upon ERAD deficiency and affects the pmr1∆ pheno-
                                   membrane protein(s) accumulates upon ERAD deficiency and affects the pmr1∆ phenotype.
                                    type. Furthermore, there may be many kinds of substrates that accumulate in ERAD-de-
                                   Furthermore, there may be many kinds of substrates that accumulate in ERAD-defective
                                    fective cells and affect the phenotype of pmr1∆ cells. One candidate is Cdc1, which sup-
                                   cells and affect the phenotype of pmr1∆ cells. One candidate is Cdc1, which suppresses
                                    presses
                                   the EGTAthe    EGTA sensitivity
                                               sensitivity of pmr1∆of   pmr1∆
                                                                      cells whencells when overexpressed
                                                                                  overexpressed               [85,119–121].
                                                                                                    [85,119–121].           Consist-
                                                                                                                   Consistently,  N-
                                    ently, N-terminal  hemagglutinin-tagged     Cdc1  is stabilized  in the Hrd3  mutant
                                   terminal hemagglutinin-tagged Cdc1 is stabilized in the Hrd3 mutant [85]. However,     [85]. How-
                                                                                                                                   a
                                    ever, a recent study  showed   that Cdc1  localizes to cis and  medial  Golgi when
                                   recent study showed that Cdc1 localizes to cis and medial Golgi when tagged with the tagged   with
                                    the fluorescent
                                   fluorescent  mNeonmNeon    protein
                                                        protein        at or
                                                                at its N- itsC-terminus
                                                                              N- or C-terminus     [94], although
                                                                                          [94], although           a previous
                                                                                                          a previous            study
                                                                                                                     study showed
                                   that Cdc1 localizes to the ER when tagged with the myc-epitope at the N-terminus [120].
                                   The reason for these discrepancies is not clear; however, the removal of EtN-P from Man 1
                                   would be a critical step for the quality control of GPI-anchored proteins before they are
                                   delivered to the plasma membrane. Nonetheless, it will be essential to test the localization
                                   and stability of a purely endogenous untagged version of Cdc1 to fully understand the
                                   observed phenomena.
Int. J. Mol. Sci. 2021, 22, 1061                                                                                                    9 of 14

                                   6. Conclusions
                                        Unidentified endogenous ERAD substrates are key to understanding the potential
                                   relationship between ERAD and the biosynthesis of GPI-anchored proteins. The simple
                                   genetic interaction reported by systematic studies implies the existence of an endogenous
                                   ERAD substrate that may affect the phenotype of cells defective in GPI biosynthesis.
                                   Mammals may express a putative positive regulator of GPI biosynthesis whose stability is
                                   regulated by ERAD. The rescue of the EGTA sensitivity of pmr1∆ cells by deletion of ERAD
                                   components could also be explained by the accumulation of endogenous substrates. This
                                   suggests that ERAD could control manganese homeostasis, which is critical for the ER exit
                                   of GPI-anchored proteins. Current data indicate that the role of ERAD is not limited to the
                                   degradation of misfolded proteins and may play a critical role in the regulation of cellular
                                   phenomena, even under normal growth conditions. The identification of an endogenous
                                   substrate of Hrd1, Doa10, as well as the dedicated ubiquitin ligases in mammals [15],
                                   would be important to fully understand the physiological roles of ERAD.

                                   Supplementary Materials: Supplementary materials can be found at https://www.mdpi.com/1422
                                   -0067/22/3/1061/s1.
                                   Funding: This work was supported by the Toray Science Foundation, the Toyoaki Scholarship
                                   Foundation, and JSPS KAKENHI, Grant Numbers 15K18503, 18K19306, and 19H02923.
                                   Institutional Review Board Statement: Not applicable.
                                   Informed Consent Statement: Not applicable.
                                   Data Availability Statement: Data is contained within the article or supplementary material.
                                   Acknowledgments: I would like to thank Takumi Kamura and Maiko Shimizu for the initial study
                                   of this project.
                                   Conflicts of Interest: The author declares no conflict of interest.

                                   Abbreviations

                                   ERAD        Endoplasmic reticulum-associated degradation
                                   EGTA        Ethylene glycol-bis(β-aminoethyl ether)-N,N,N 0 ,N 0 -tetra-acetic acid
                                   GPI         Glycosylphosphatidylinositol
                                   EtN-P       Ethanolamine phosphate
                                   Man         Mannose
                                   GlcN        Glucosamine
                                   GlcNAc      N-Acetylglucosamine
                                   UPR         Unfolded protein response
                                   prERAD      Pre-insertional ERAD

References
1.    Rüdiger, S.; Buchberger, A.; Bukau, B. Interaction of Hsp70 chaperones with substrates. Nat. Struct. Mol. Biol. 1997, 4, 342–349.
      [CrossRef] [PubMed]
2.    Rutkowski, D.; Kaufman, R.J. A trip to the ER: Coping with stress. Trends Cell Biol. 2004, 14, 20–28. [CrossRef] [PubMed]
3.    Ron, D.; Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat. Rev. Mol. Cell Biol. 2007, 8,
      519–529. [CrossRef] [PubMed]
4.    Vembar, S.S.; Brodsky, J.L. One step at a time: Endoplasmic reticulum-associated degradation. Nat. Rev. Mol. Cell Biol. 2008, 9,
      944–957. [CrossRef]
5.    Smith, M.H.; Ploegh, H.L.; Weissman, J.S. Road to Ruin: Targeting Proteins for Degradation in the Endoplasmic Reticulum.
      Science 2011, 334, 1086–1090. [CrossRef]
6.    Ruggiano, A.; Foresti, O.; Carvalho, P. Quality control: ER-associated degradation: Protein quality control and beyond. J. Cell Biol.
      2014, 204, 869–879. [CrossRef]
7.    Oikonomou, C.; Hendershot, L.M. Disposing of misfolded ER proteins: A troubled substrate’s way out of the ER. Mol. Cell
      Endocrinol. 2020, 500, 110630. [CrossRef]
8.    Sun, Z.; Brodsky, J.L. Protein quality control in the secretory pathway. J. Cell Biol. 2019, 218, 3171–3187. [CrossRef]
Int. J. Mol. Sci. 2021, 22, 1061                                                                                                    10 of 14

9.    Mehrtash, A.B.; Hochstrasser, M. Ubiquitin-dependent protein degradation at the endoplasmic reticulum and nuclear envelope.
      Semin. Cell Dev. Biol. 2019, 93, 111–124. [CrossRef]
10.   Berner, N.; Reutter, K.-R.; Wolf, D.H. Protein Quality Control of the Endoplasmic Reticulum and Ubiquitin–Proteasome-Triggered
      Degradation of Aberrant Proteins: Yeast Pioneers the Path. Annu. Rev. Biochem. 2018, 87, 751–782. [CrossRef]
11.   Christianson, J.C.; Ye, Y. Cleaning up in the endoplasmic reticulum: Ubiquitin in charge. Nat. Struct. Mol. Biol. 2014, 21, 325–335.
      [CrossRef] [PubMed]
12.   Wu, X.; Rapoport, T.A. Mechanistic insights into ER-associated protein degradation. Curr. Opin. Cell Biol. 2018, 53, 22–28.
      [CrossRef] [PubMed]
13.   Nakatsukasa, K.; Brodsky, J.L. The Recognition and Retrotranslocation of Misfolded Proteins from the Endoplasmic Reticulum.
      Traffic 2008, 9, 861–870. [CrossRef] [PubMed]
14.   Nakatsukasa, K.; Okumura, F.; Kamura, T. Proteolytic regulation of metabolic enzymes by E3 ubiquitin ligase complexes: Lessons
      from yeast. Crit. Rev. Biochem. Mol. Biol. 2015, 50, 489–502. [CrossRef] [PubMed]
15.   Qi, L.; Tsai, B.; Arvan, P. New Insights into the Physiological Role of Endoplasmic Reticulum-Associated Degradation. Trends Cell
      Biol. 2017, 27, 430–440. [CrossRef]
16.   Adle, D.J.; Wei, W.; Smith, N.; Bies, J.J.; Lee, J. Cadmium-mediated rescue from ER-associated degradation induces expression of
      its exporter. Proc. Natl. Acad. Sci. USA 2009, 106, 10189–10194. [CrossRef]
17.   Hampton, R.Y.; Gardner, R.G.; Rine, J. Role of 26S proteasome and HRD genes in the degradation of 3-hydroxy-3-methylglutaryl-
      CoA reductase, an integral endoplasmic reticulum membrane protein. Mol. Biol. Cell 1996, 7, 2029–2044. [CrossRef]
18.   Rape, M.; Hoppe, T.; Gorr, I.; Kalocay, M.; Richly, H.; Jentsch, S. Mobilization of Processed, Membrane-Tethered SPT23
      Transcription Factor by CDC48UFD1/NPL4, a Ubiquitin-Selective Chaperone. Cell 2001, 107, 667–677. [CrossRef]
19.   Foresti, O.; Ruggiano, A.; Hannibal-Bach, H.K.; Ejsing, C.S.; Carvalho, P. Sterol homeostasis requires regulated degradation of
      squalene monooxygenase by the ubiquitin ligase Doa10/Teb4. eLife 2013, 2, e00953. [CrossRef]
20.   Wu, X.; Siggel, M.; Ovchinnikov, S.; Mi, W.; Svetlov, V.; Nudler, E.; Liao, M.; Hummer, G.; Rapoport, T.A. Structural basis of
      ER-associated protein degradation mediated by the Hrd1 ubiquitin ligase complex. Science 2020, 368, eaaz2449. [CrossRef]
21.   Knop, M.; Finger, A.; Braun, T.; Hellmuth, K.; Wolf, D.H. Der1, a novel protein specifically required for endoplasmic reticulum
      degradation in yeast. EMBO J. 1996, 15, 753–763. [CrossRef] [PubMed]
22.   Bordallo, J.; Plemper, R.K.; Finger, A.; Wolf, D.H. Der3p/Hrd1p Is Required for Endoplasmic Reticulum-associated Degradation
      of Misfolded Lumenal and Integral Membrane Proteins. Mol. Biol. Cell 1998, 9, 209–222. [CrossRef] [PubMed]
23.   Bays, N.W.; Gardner, R.G.; Seelig, L.P.; Joazeiro, C.A.; Hampton, R.Y. Hrd1p/Der3p is a membrane-anchored ubiquitin ligase
      required for ER-associated degradation. Nat. Cell Biol. 2001, 3, 24–29. [CrossRef] [PubMed]
24.   Gauss, R.; Jarosch, E.; Sommer, T.; Hirsch, C. A complex of Yos9p and the HRD ligase integrates endoplasmic reticulum quality
      control into the degradation machinery. Nat. Cell Biol. 2006, 8, 849–854. [CrossRef]
25.   Carvalho, P.; Goder, V.; Rapoport, T.A. Distinct Ubiquitin-Ligase Complexes Define Convergent Pathways for the Degradation of
      ER Proteins. Cell 2006, 126, 361–373. [CrossRef]
26.   Kanehara, K.; Xie, W.; Ng, D.T.W. Modularity of the Hrd1 ERAD complex underlies its diverse client range. J. Cell Biol. 2010, 188,
      707–716. [CrossRef]
27.   Mehnert, M.; Sommer, T.; Jarosch, E. Der1 promotes movement of misfolded proteins through the endoplasmic reticulum
      membrane. Nat. Cell Biol. 2014, 16, 77–86. [CrossRef]
28.   Gauss, R.; Sommer, T.; Jarosch, E. The Hrd1p ligase complex forms a linchpin between ER-lumenal substrate selection and Cdc48p
      recruitment. EMBO J. 2006, 25, 1827–1835. [CrossRef]
29.   Bhamidipati, A.; Denic, V.; Quan, E.M.; Weissman, J.S. Exploration of the Topological Requirements of ERAD Identifies Yos9p as
      a Lectin Sensor of Misfolded Glycoproteins in the ER Lumen. Mol. Cell 2005, 19, 741–751. [CrossRef]
30.   Szathmary, R.; Bielmann, R.; Nita-Lazar, M.; Burda, P.; Jakob, C.A. Yos9 Protein Is Essential for Degradation of Misfolded
      Glycoproteins and May Function as Lectin in ERAD. Mol. Cell 2005, 19, 765–775. [CrossRef]
31.   Quan, E.M.; Kamiya, Y.; Kamiya, D.; Denic, V.; Weibezahn, J.; Kato, K.; Weissman, J.S. Defining the Glycan Destruction Signal for
      Endoplasmic Reticulum-Associated Degradation. Mol. Cell 2008, 32, 870–877. [CrossRef] [PubMed]
32.   Jakob, C.A.; Bodmer, D.; Spirig, U.; Bättig, P.; Marcil, A.; Dignard, D.; Bergeron, J.J.; Thomas, D.Y.; Aebi, M. Htm1p, a mannosidase-
      like protein, is involved in glycoprotein degradation in yeast. EMBO Rep. 2001, 2, 423–430. [CrossRef] [PubMed]
33.   Nakatsukasa, K.; Nishikawa, S.-I.; Hosokawa, N.; Nagata, K.; Endo, T. Mnl1p, an α-Mannosidase-like Protein in YeastSaccha-
      romyces cerevisiae, Is Required for Endoplasmic Reticulum-associated Degradation of Glycoproteins. J. Biol. Chem. 2001, 276,
      8635–8638. [CrossRef] [PubMed]
34.   Xie, W.; Kanehara, K.; Sayeed, A.; Ng, D.T.W. Intrinsic Conformational Determinants Signal Protein Misfolding to the Hrd1/Htm1
      Endoplasmic Reticulum–associated Degradation System. Mol. Biol. Cell 2009, 20, 3317–3329. [CrossRef] [PubMed]
35.   Vashistha, N.; Neal, S.E.; Singh, A.; Carroll, S.M.; Hampton, R.Y. Direct and essential function for Hrd3 in ER-associated
      degradation. Proc. Natl. Acad. Sci. USA 2016, 113, 5934–5939. [CrossRef]
36.   Horn, S.C.; Hanna, J.; Hirsch, C.; Volkwein, C.; Schütz, A.; Heinemann, U.; Sommer, T.; Jarosch, E. Usa1 Functions as a Scaffold of
      the HRD-Ubiquitin Ligase. Mol. Cell 2009, 36, 782–793. [CrossRef]
37.   Biederer, T.; Volkwein, C.; Sommer, T. Role of Cue1p in Ubiquitination and Degradation at the ER Surface. Science 1997, 278,
      1806–1809. [CrossRef]
Int. J. Mol. Sci. 2021, 22, 1061                                                                                                   11 of 14

38.   Schuberth, C.; Buchberger, A. Membrane-bound Ubx2 recruits Cdc48 to ubiquitin ligases and their substrates to ensure efficient
      ER-associated protein degradation. Nat. Cell Biol. 2005, 7, 999–1006. [CrossRef]
39.   Neuber, O.; Jarosch, E.; Volkwein, C.; Walter, J.; Sommer, T. Ubx2 links the Cdc48 complex to ER-associated protein degradation.
      Nat. Cell Biol. 2005, 7, 993–998. [CrossRef]
40.   Nakatsukasa, K.; Brodsky, J.L.; Kamura, T. A stalled retrotranslocation complex reveals physical linkage between substrate
      recognition and proteasomal degradation during ER-associated degradation. Mol. Biol. Cell 2013, 24, 1765–1775. [CrossRef]
41.   Sato, B.K.; Schulz, D.; Do, P.H.; Hampton, R.Y. Misfolded Membrane Proteins Are Specifically Recognized by the Transmembrane
      Domain of the Hrd1p Ubiquitin Ligase. Mol. Cell 2009, 34, 212–222. [CrossRef] [PubMed]
42.   Neal, S.; Jaeger, P.A.; Duttke, S.H.; Benner, C.; Glass, C.K.; Ideker, T.; Hampton, R.Y. The Dfm1 Derlin Is Required for ERAD
      Retrotranslocation of Integral Membrane Proteins. Mol. Cell 2018, 69, 306–320.e4. [CrossRef] [PubMed]
43.   Neal, S.E.; Syau, D.; Nejatfard, A.; Nadeau, S.; Hampton, R.Y. HRD Complex Self-Remodeling Enables a Novel Route of
      Membrane Protein Retrotranslocation. iScience 2020, 23, 101493. [CrossRef] [PubMed]
44.   Kreft, S.G.; Wang, L.; Hochstrasser, M. Membrane Topology of the Yeast Endoplasmic Reticulum-localized Ubiquitin Ligase
      Doa10 and Comparison with Its Human Ortholog TEB4 (MARCH-VI). J. Biol. Chem. 2006, 281, 4646–4653. [CrossRef]
45.   Huyer, G.; Piluek, W.F.; Fansler, Z.; Kreft, S.G.; Hochstrasser, M.; Brodsky, J.L.; Michaelis, S. Distinct machinery is required in
      Saccharomyces cerevisiae for the endoplasmic reticulum-associated degradation of a multispanning membrane protein and a
      soluble luminal protein. J. Biol. Chem. 2004, 279, 38369–38378. [CrossRef]
46.   Gnann, A.; Riordan, J.R.; Wolf, D.H. Cystic Fibrosis Transmembrane Conductance Regulator Degradation Depends on the Lectins
      Htm1p/EDEM and the Cdc48 Protein Complex in Yeast. Mol. Biol. Cell 2004, 15, 4125–4135. [CrossRef]
47.   Ravid, T.; Kreft, S.G.; Hochstrasser, M. Membrane and soluble substrates of the Doa10 ubiquitin ligase are degraded by distinct
      pathways. EMBO J. 2006, 25, 533–543. [CrossRef]
48.   Vashist, S.; Ng, D.T. Misfolded proteins are sorted by a sequential checkpoint mechanism of ER quality control. J. Cell Biol. 2004,
      165, 41–52. [CrossRef]
49.   Furth, N.; Gertman, O.; Shiber, A.; Alfassy, O.S.; Cohen, I.; Rosenberg, M.M.; Doron, N.K.; Friedler, A.; Ravid, T. Exposure of
      bipartite hydrophobic signal triggers nuclear quality control of Ndc10 at the endoplasmic reticulum/nuclear envelope. Mol. Biol.
      Cell 2011, 22, 4726–4739. [CrossRef]
50.   Swanson, R.; Locher, M.; Hochstrasser, M. A conserved ubiquitin ligase of the nuclear envelope/endoplasmic reticulum that
      functions in both ER-associated and Matalpha2 repressor degradation. Genes Dev. 2001, 15, 2660–2674. [CrossRef]
51.   Habeck, G.; Ebner, F.A.; Shimada-Kreft, H.; Kreft, S.G. The yeast ERAD-C ubiquitin ligase Doa10 recognizes an intramembrane
      degron. J. Cell Biol. 2015, 209, 261–273. [CrossRef] [PubMed]
52.   Schmidt, C.C.; Vasic, V.; Stein, A. Doa10 is a membrane protein retrotranslocase in ER-associated protein degradation. eLife 2020,
      9, e56945. [CrossRef] [PubMed]
53.   Nakatsukasa, K.; Huyer, G.; Michaelis, S.; Brodsky, J.L. Dissecting the ER-Associated Degradation of a Misfolded Polytopic
      Membrane Protein. Cell 2008, 132, 101–112. [CrossRef] [PubMed]
54.   Han, S.; Liu, Y.; Chang, A. Cytoplasmic Hsp70 Promotes Ubiquitination for Endoplasmic Reticulum-associated Degradation of a
      Misfolded Mutant of the Yeast Plasma Membrane ATPase, PMA1. J. Biol. Chem. 2007, 282, 26140–26149. [CrossRef]
55.   Metzger, M.B.; Maurer, M.J.; Dancy, B.M.; Michaelis, S. Degradation of a Cytosolic Protein Requires Endoplasmic Reticulum-
      associated Degradation Machinery. J. Biol. Chem. 2008, 283, 32302–32316. [CrossRef]
56.   Weber, A.; Cohen, I.; Popp, O.; Dittmar, G.; Reiss, Y.; Sommer, T.; Ravid, T.; Jarosch, E. Sequential Poly-ubiquitylation by
      Specialized Conjugating Enzymes Expands the Versatility of a Quality Control Ubiquitin Ligase. Mol. Cell 2016, 63, 827–839.
      [CrossRef]
57.   Nakatsukasa, K.; Kamura, T. Subcellular Fractionation Analysis of the Extraction of Ubiquitinated Polytopic Membrane Substrate
      during ER-Associated Degradation. PLoS ONE 2016, 11, e0148327. [CrossRef]
58.   Walter, J.; Urban, J.; Volkwein, C.; Sommer, T. Sec61p-independent degradation of the tail-anchored ER membrane protein Ubc6p.
      EMBO J. 2001, 20, 3124–3131. [CrossRef]
59.   Pittet, M.; Conzelmann, A. Biosynthesis and function of GPI proteins in the yeast Saccharomyces cerevisiae. Biochim. Et Biophys.
      Acta (BBA)-Mol. Cell Biol. Lipids 2007, 1771, 405–420. [CrossRef]
60.   Leidich, S.D.; Kostova, Z.; Latek, R.R.; Costello, L.C.; Drapp, D.A.; Gray, W.; Fassler, J.S.; Orlean, P. Temperature-sensitive Yeast
      GPI Anchoring Mutants gpi2 and gpi3 Are Defective in the Synthesis of N-Acetylglucosaminyl Phosphatidylinositol. J. Biol.
      Chem. 1995, 270, 13029–13035. [CrossRef]
61.   Yan, B.C.; Westfall, B.A.; Orlean, P. Ynl038wp (Gpi15p) is theSaccharomyces cerevisiae homologue of human Pig-Hp and
      participates in the first step in glycosylphosphatidylinositol assembly. Yeast 2001, 18, 1383–1389. [CrossRef] [PubMed]
62.   Newman, H.A.; Romeo, M.J.; Lewis, S.E.; Yan, B.C.; Orlean, P.; Levin, D.E. Gpi19, the Saccharomyces cerevisiae Homologue of
      Mammalian PIG-P, Is a Subunit of the Initial Enzyme for Glycosylphosphatidylinositol Anchor Biosynthesis. Eukaryot. Cell 2005,
      4, 1801–1807. [CrossRef] [PubMed]
63.   Orlean, P.; Menon, A.K. Thematic review series: Lipid Posttranslational Modifications.GPI anchoring of protein in yeast and
      mammalian cells, or: How we learned to stop worrying and love glycophospholipids. J. Lipid Res. 2007, 48, 993–1011. [CrossRef]
      [PubMed]
Int. J. Mol. Sci. 2021, 22, 1061                                                                                                   12 of 14

64.   Maeda, Y.; Watanabe, R.; Harris, C.L.; Hong, Y.; Ohishi, K.; Kinoshita, K.; Kinoshita, T. PIG-M transfers the first mannose to
      glycosylphosphatidylinositol on the lumenal side of the ER. EMBO J. 2001, 20, 250–261. [CrossRef]
65.   Kang, J.Y.; Hong, Y.; Ashida, H.; Shishioh, N.; Murakami, Y.; Morita, Y.S.; Maeda, Y.; Kinoshita, T. PIG-V Involved in Transferring
      the Second Mannose in Glycosylphosphatidylinositol. J. Biol. Chem. 2005, 280, 9489–9497. [CrossRef]
66.   Gaynor, E.C.; Mondésert, G.; Grimme, S.J.; Reed, S.I.; Orlean, P.; Emr, S.D. MCD4Encodes a Conserved Endoplasmic Reticulum
      Membrane Protein Essential for Glycosylphosphatidylinositol Anchor Synthesis in Yeast. Mol. Biol. Cell 1999, 10, 627–648.
      [CrossRef]
67.   Sütterlin, C.; Escribano, M.V.; Gerold, P.; Maeda, Y.; Mazón, M.J.; Kinoshita, T.; Schwarz, R.T.; Riezman, H. Saccharomyces
      cerevisiae GPI10, the functional homologue of human PIG-B, is required for glycosylphosphatidylinositol-anchor synthesis.
      Biochem. J. 1998, 332, 153–159. [CrossRef]
68.   Flury, I.; Benachour, A.; Conzelmann, A. YLL031c Belongs to a Novel Family of Membrane Proteins Involved in the Transfer
      of Ethanolaminephosphate onto the Core Structure of Glycosylphosphatidylinositol Anchors in Yeast. J. Biol. Chem. 2000, 275,
      24458–24465. [CrossRef]
69.   Grimme, S.J.; Westfall, B.A.; Wiedman, J.M.; Taron, C.H.; Orlean, P. The Essential Smp3 Protein Is Required for Addition of the
      Side-branching Fourth Mannose during Assembly of Yeast Glycosylphosphatidylinositols. J. Biol. Chem. 2001, 276, 27731–27739.
      [CrossRef]
70.   Benachour, A.; Sipos, G.; Flury, I.; Reggiori, F.; Canivenc-Gansel, E.; Vionnet, C.; Conzelmann, A.; Benghezal, M. Deletion of GPI7,
      a Yeast Gene Required for Addition of a Side Chain to the Glycosylphosphatidylinositol (GPI) Core Structure, Affects GPI Protein
      Transport, Remodeling, and Cell Wall Integrity. J. Biol. Chem. 1999, 274, 15251–15261. [CrossRef]
71.   Imhof, I.; Flury, I.; Vionnet, C.; Roubaty, C.; Egger, D.; Conzelmann, A. Glycosylphosphatidylinositol (GPI) Proteins ofSaccha-
      romyces cerevisiaeContain Ethanolamine Phosphate Groups on the α1,4-linked Mannose of the GPI Anchor. J. Biol. Chem. 2004,
      279, 19614–19627. [CrossRef] [PubMed]
72.   Chen, R.; Knez, J.J.; Merrick, W.C.; Medof, M.E. Comparative efficiencies of C-terminal signals of native glycophosphatidylinositol
      (GPI)-anchored proproteins in conferring GPI-anchoring. J. Cell. Biochem. 2002, 84, 68–83. [CrossRef] [PubMed]
73.   Ohishi, K.; Inoue, N.; Kinoshita, T. PIG-S and PIG-T, essential for GPI anchor attachment to proteins, form a complex with GAA1
      and GPI8. EMBO J. 2001, 20, 4088–4098. [CrossRef]
74.   Benghezal, M.; Benachour, A.; Rusconi, S.; Aebi, M.; Conzelmann, A. Yeast Gpi8p is essential for GPI anchor attachment onto
      proteins. EMBO J. 1996, 15, 6575–6583. [CrossRef] [PubMed]
75.   Fraering, P.; Imhof, I.; Meyer, U.; Strub, J.-M.; Van Dorsselaer, A.; Vionnet, C.; Conzelmann, A. The GPI Transamidase Complex of
      Saccharomyces cerevisiae Contains Gaa1p, Gpi8p, and Gpi16p. Mol. Biol. Cell 2001, 12, 3295–3306. [CrossRef]
76.   Hamburger, D.; Egerton, M.; Riezman, H. Yeast Gaa1p is required for attachment of a completed GPI anchor onto proteins. J. Cell
      Biol. 1995, 129, 629–639. [CrossRef]
77.   Hong, Y.; Ohishi, K.; Kang, J.Y.; Tanaka, S.; Inoue, N.; Nishimura, J.-I.; Maeda, Y.; Kinoshita, T. Human PIG-U and Yeast Cdc91p
      Are the Fifth Subunit of GPI Transamidase That Attaches GPI-Anchors to Proteins. Mol. Biol. Cell 2003, 14, 1780–1789. [CrossRef]
78.   Meyer, U.; Benghezal, M.; Imhof, I.; Conzelmann, A. Active Site Determination of Gpi8p, a Caspase-Related Enzyme Required for
      Glycosylphosphatidylinositol Anchor Addition to Proteins†. Biochemistry 2000, 39, 3461–3471. [CrossRef]
79.   Ohishi, K.; Inoue, N.; Maeda, Y.; Takeda, J.; Riezman, H.; Kinoshita, T. Gaa1p and Gpi8p Are Components of a Glycosylphos-
      phatidylinositol (GPI) Transamidase That Mediates Attachment of GPI to Proteins. Mol. Biol. Cell 2000, 11, 1523–1533. [CrossRef]
80.   Kang, X.; Szallies, A.; Rawer, M.; Echner, H.; Duszenko, M. GPI anchor transamidase of Trypanosoma brucei: In vitro assay of the
      recombinant protein and VSG anchor exchange. J. Cell Sci. 2002, 115, 2529–2539.
81.   Tanaka, S.; Maeda, Y.; Tashima, Y.; Kinoshita, T. Inositol Deacylation of Glycosylphosphatidylinositol-anchored Proteins Is
      Mediated by Mammalian PGAP1 and Yeast Bst1p. J. Biol. Chem. 2004, 279, 14256–14263. [CrossRef] [PubMed]
82.   Fujita, M.; Umemura, M.; Yoko-o, T.; Jigami, Y. PER1 Is Required for GPI-Phospholipase A2 Activity and Involved in Lipid
      Remodeling of GPI-anchored Proteins. Mol. Biol. Cell 2006, 17, 5253–5264. [CrossRef] [PubMed]
83.   Bosson, R.; Jaquenoud, M.; Conzelmann, A. GUP1 of Saccharomyces cerevisiae Encodes an O-Acyltransferase Involved in
      Remodeling of the GPI Anchor. Mol. Biol. Cell 2006, 17, 2636–2645. [CrossRef] [PubMed]
84.   Umemura, M.; Fujita, M.; Yoko-o, T.; Fukamizu, A.; Jigami, Y. Saccharomyces cerevisiae CWH43Is Involved in the Remodeling of
      the Lipid Moiety of GPI Anchors to Ceramides. Mol. Biol. Cell 2007, 18, 4304–4316. [CrossRef]
85.   Vazquez, H.M.; Vionnet, C.; Roubaty, C.; Conzelmann, A. Cdc1 removes the ethanolamine phosphate of the first mannose of
      GPI anchors and thereby facilitates the integration of GPI proteins into the yeast cell wall. Mol. Biol. Cell 2014, 25, 3375–3388.
      [CrossRef] [PubMed]
86.   Manzano-Lopez, J.; Perez-Linero, A.M.; Aguilera-Romero, A.; Martin, M.E.; Okano, T.; Silva, D.V.; Seeberger, P.H.; Riezman, H.;
      Funato, K.; Goder, V.; et al. COPII Coat Composition Is Actively Regulated by Luminal Cargo Maturation. Curr. Biol. 2015, 25,
      152–162. [CrossRef]
87.   Fujita, M.; Maeda, Y.; Ra, M.; Yamaguchi, Y.; Taguchi, R.; Kinoshita, T. GPI Glycan Remodeling by PGAP5 Regulates Transport of
      GPI-Anchored Proteins from the ER to the Golgi. Cell 2009, 139, 352–365. [CrossRef]
88.   Fujita, M.; Yoko-o, T.; Jigami, Y. Inositol Deacylation by Bst1p Is Required for the Quality Control of Glycosylphosphatidylinositol-
      anchored Proteins. Mol. Biol. Cell 2006, 17, 834–850. [CrossRef]
You can also read